You are on page 1of 12

Montrose et al.

Allergy Asthma Clin Immunol (2017) 13:12 Allergy, Asthma & Clinical Immunology
DOI 10.1186/s13223-017-0187-8

RESEARCH Open Access

Dietary intake is associated


with respiratory health outcomes and DNA
methylation in children with asthma
L. Montrose1, T. J. Ward2, E. O. Semmens2, Y. H. Cho2, B. Brown3 and C. W. Noonan2*

Abstract 
Background:  Asthma is an increasingly common chronic disease among children, and data point toward a complex
mechanism involving genetic, environmental and epigenetic factors. Epigenetic modifications such as DNA hypo- or
hyper-methylation have been shown to occur in response to environmental exposures including dietary nutrients.
Methods:  Within the context of the asthma randomized trial of indoor wood smoke (ARTIS) study, we investigated
relationships between diet, asthma health measures, and DNA methylation. Asthma health measures included a
quality of life instrument, diurnal peak flow variability (dPFV) and forced expiratory volume in the first second (FEV1).
Dietary intake was assessed with a food frequency questionnaire. Methylation levels of LINE-1 repetitive element and
two promoter CpG sites for interferon gamma (IFNγ, -186 and -54) from buccal cell DNA were measured using pyrose-
quencing assays.
Results:  Data were collected on 32 children with asthma living in western Montana who were recruited to the ARTIS
study. Selenium and several methyl donor dietary nutrients were positively associated with the asthma quality of life
measure. Intake of methyl donating nutrients including folate was positively associated LINE-1 methylation and nega-
tively associated with IFNγ CpG-186. Higher levels of LINE-1 methylation were associated with greater dPFV.
Conclusion:  We identified several nutrients that were associated with improved quality of life measures among
children with asthma. The IFNγ promoter CpG site -186 but not -54 was associated with the intake of selected dietary
nutrients. However, in this small population of children with asthma, the IFNγ promoter CpG sites were not associ-
ated with respiratory health measures so it remains unclear through which epigenetic mechanism these nutrients
are impacting the quality of life measure. These findings add to the evidence that dietary nutrients, particularly foods
containing methyl donors, may be important for epigenetic regulation as it pertains to the control of asthma.
Trial registration ClincialTrials.gov NCT00807183. Registered 10 December 2008
Keywords:  Asthma, Methylation, Spirometry, Diet, Nutrition, Children, Epigenetics, Quality of life

Background countries [2]. Epidemiological studies suggest that die-


Asthma is an environmentally triggered disease that tary patterns are linked to the risk of developing asthma,
affects nearly 26 million people in the United States [1]. however, the evidence from longitudinal birth cohorts
Dietary intake represents a modifiable environmental has not clearly defined the importance of specific nutri-
exposure that could partially explain the current burden ents or fully elucidated the mechanistic pathways linking
of chronic disease, including asthma, in industrialized diet to chronic respiratory disease. Further, there have
been few studies aimed at determining if nutrient intake
contributes to asthma control in children. One potential
*Correspondence: curtis.noonan@mso.umt.edu mechanism whereby dietary intake affects respiratory
2
Center for Environmental Health Sciences, University of Montana, 32 health in children is through epigenetic modulation of
Campus Drive‑159 Skaggs, Missoula, MT 59812, USA
Full list of author information is available at the end of the article
immunoregulatory cytokines.

© The Author(s) 2017. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium,
provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license,
and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/
publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 2 of 12

Significant observational data suggests that dietary adolescent, or adult). Nevertheless, human and mouse
status and intake of particular nutrients can affect res- data indicate several dietary nutrients play a role in epi-
piratory health outcomes. Several recent studies have genetic mechanisms [24], thus it is possible that nutri-
suggested that some dietary nutrients may be protective ent intake is related to asthma pathogenesis through the
for respiratory health [3–12]. A recent review concluded epigenetic regulation of key genes. Asthma is pheno-
that dietary intake in utero and throughout the lifecourse typically characterized by a shift toward type 2 T helper
can influence respiratory health status, however defini- (Th2) polarization and consequently type 1 T helper
tively assessing causal relationships in human studies is a (Th1) cell cytokines such as interferon gamma (IFNγ)
major challenge [13]. A cross-sectional study by Berthon play a critical role as counter regulators in the allergic
et al. showed that among asthmatics, a high fat diet was asthma pathway [25, 26]. For example, in a follow-up
associated with increased airway eosinophilic inflamma- study of adults recruited as children with a history of
tion, and low fiber intake was associated with poor lung wheeze, those with persistent asthma were compared to
function [14]. Supplementation of dietary folic acid has those with resolved asthma to characterize the Th1/Th2
been successful in the prevention of neural tube defects response following exposure to house dust mite allergen
in the United States. However, longitudinal cohort stud- [27]. Smart et al. found that those with persistent asthma
ies have produced mixed results regarding maternal had much weaker Th1 responses and concluded that a
folic acid supplementation and asthma development [15, measured decrease in IFNγ production in this group
16]. Antioxidants like selenium may play a role in res- could be a major factor underpinning the presence of
piratory health through systemic reduction of oxidative severe and chronic asthma symptoms. Meng et al. inves-
stress [17]. In a mouse model of allergic airway disease, a tigated the effect of diet on IFNγ production in humans
combinatory therapeutic that included selenium attenu- and showed that cells extracted and purified from non-
ated the physiologic airway damage that is typical of this asthmatic adults produced differential amounts of IFNγ
model [18]. [28]. Interestingly, Meng found that the amounts of IFNγ
The rapidly evolving field of epigenetics has emerged were associated with intake of specific dietary variables
as an appealing potential mechanistic bridge that could and predicted upper respiratory tract infection incidence.
link environmental exposures to the development of Finally, a series of studies using either a ragweed or dust
asthma or the exacerbation of asthma-related symptoms mite-sensitized mouse model of asthma showed that pre-
[19]. The exact toxicoepigenetic mechanisms are far from treatment with a DNA adjuvant known to result in Th1
elucidated, but landmark studies using the agouti mouse biased immune status with marked overproduction of
model have provided solid evidence that environmental IFNγ resulted in an ameliorated lung inflammatory phe-
exposures can affect phenotype through alterations in notype [29, 30]. Thus IFNγ is a relevant candidate gene
DNA methylation patterns [20]. Understanding how and that plausibly exists in the mechanistic pathway linking
when these mechanisms can impact asthma pathogenesis dietary intake to respiratory health via epigenetic regula-
is paramount. In a mouse model of allergic airway dis- tion of the Th1/Th2 cytokine balance.
ease, in utero dietary intake of methyl donating nutrients Poor asthma control is associated with school absences,
was associated with an enhanced disease phenotype as higher health care costs and worse long-term health out-
well as aberrant hypermethylation of runt-related tran- comes. An understanding of the association between
scription factor 3 (Runx3), a gene known to suppress a child’s recent dietary history and respiratory health
allergic airway disease [21]. Although the perinatal expo- measures could lead to important intervention strategies
sure window may be especially important, data also sug- to improve outcomes among children with asthma. In
gest that environmental exposures could impact health this study we aimed to evaluate the relationship between
via epigenetic mechanisms throughout the lifecourse. a priori selected nutrients and asthma health. Although
In humans, the production of regulatory T cells (Tregs), the link between current dietary status and asthma
which are known to suppress immune responses, is con- health is not clear, evidence suggests a potential role for
trolled by transcription factor forkhead box p3 (FOXP3) an epigenetic mechanism. In addition to a measure of
[22]. Nadeau et  al. demonstrated that patients with global gene methylation, IFNγ was chosen as a candidate
asthma in a polluted environment had a hypermethylated gene because of its well-established role in the Th1/Th2
FOXP3 locus profile which was associated with impaired balance.
Treg function relative to patients with asthma in a less
polluted area [23]. Methods
The relationship between dietary intake and epige- Study overview
netic modifications is complex and compounded by sen- Participants were recruited from the asthma randomized
sitivity to timing of exposure (e.g. prenatal, postnatal, trial of indoor wood smoke (ARTIS) study. The rationale
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 3 of 12

and methods for the ARTIS study have been described Health outcome measures collected in parent study
previously [31–33]. The ARTIS study included 114 chil- The pediatric asthma quality of life questionnaire
dren with asthma (ages 6–17) from 97 homes in Mon- (PAQLQ) is a 23-item asthma-specific battery which
tana, Idaho, and Alaska. This parent study was designed provides domain scores for symptoms (10 items), activ-
to test an indoor air quality intervention, and homes were ity limitation (5 items), and emotional function (8 items)
assigned to either a placebo arm or an air filter interven- [39]. The total PAQLQ score and each domain score are
tion. Two in-home data collection visits occurred in each calculated as mean scores ranging from one to seven with
of two consecutive winter periods with the intervention seven as the optimal score. The PAQLQ has been vali-
occurring between these winter periods. The subco- dated as an evaluative tool to measure within participant
hort recruited to participate in this diet and epigenetics changes over time due to treatment, and changes in this
study included 32 participants living in western Montana scale of 0.5 or more points are clinically significant [39].
who had been recruited in the final 2 years of the 5-year Using the PiKo-1  m (Ferraris Respiratory, Ayer, MA,
ARTIS study. Additional file 1: Figure S1 indicates when USA) participants performed a test twice daily, in the
spirometry measurements, buccal cells, and food fre- morning and in the evening, for a period of 2 weeks.
quency questionnaires (FFQ) were administered. For the These 2-week periods were initiated at the beginning of
purpose of the currently described study, only data that each air sampling event. For each test, the child’s parent
was collected in conjunction with a FFQ was considered. records the observation as it appears on the meter and
In Additional file 1: Figure S1, this would be visits B and these observations are later checked for accuracy against
D. Health outcomes measures included a quality of life the digital log of the instrument. The instrument records
instrument and self-monitoring of spirometry measures the best result for both peak expiratory flow (PEF) and
using a peak flow meter. Buccal cell samples were col- forced expiratory volume in one second (FEV1). Out-
lected for evaluating epigenetic markers. Anthropomet- comes from these measures include average morning
ric measures determined by trained staff using a digital PEF and FEV1, average evening PEF and FEV1, and diur-
scale and stadiometer along with the participant’s gen- nal PEF variability (dPFV).
der and date of birth were used to calculate body mass
index (BMI) percentile using the U.S. Centers for Disease Cell collection, DNA extraction, and pyrosequencing
Control and Prevention (CDC) calculator [34]. The study Buccal cells were collected from the participant’s cheek
was approved by the University of Montana Institutional by trained staff using a cytology brush and stored in Cell
Review Board. In addition to the informed consent pro- Lysis Solution (Qiagen, Valencia, CA, USA) at room tem-
cedures for the parent study, children were separately perature until all samples were collected. In compliance
assented to participate in this diet and epigenetics study with this protocol, all samples were processed within
and parents signed a parental permission and informed 24  months from the day of collection. DNA from the
consent form. buccal cells was extracted using Gentra Puregene Buccal
Cell DNA Kit (Qiagen, Valencia, CA, USA) according to
Dietary nutrient collection the manufacturer’s instructions. The quantity of the puri-
Dietary data was collected using the 2004 Block Kids fied DNA was measured using a Nanodrop spectropho-
FFQ (NutritionQuest, Berkeley, CA, USA) to charac- tometer (Thermo Scientific, Wilmington, DE) and then
terize dietary intake among participants. This instru- stored at −20  °C. DNA bisulfite treatment was carried
ment has been validated in children, ages 6–17  years out using the EZ DNA Methylation-Direct Kit (Zymo
old [35–38]. The questionnaire includes 77 food items. Research, Irvine, CA) according to the manufacturer’s
In addition to intake of standard nutrients, this instru- instruction, and stored at −20 °C. Pyrosequencing assay
ment was used to estimate intake of micronutrients that was used to measure methylation levels of LINE-1 repeti-
participate in the one-carbon metabolism pathway (i.e. tive element and the promoter region of INFγ. Briefly,
betaine, choline, folate, etc.). These nutrients are impor- 50  µg of bisulfite-modified DNA was PCR amplified by
tant for the generation of methyl groups and are there- polymerase chain reaction (PCR) using specific primers
fore potentially relevant to DNA methylation markers. (Additional file  1: Table S1) and the PyroMark PCR kit
The questionnaire was administered to each participant (Qiagen, Valencia, CA, USA). After annealing, pyrose-
by trained staff using serving size visual aides, and par- quencing was conducted using a Pyromark Q96 MD
ents were asked to assist their child with portion size rec- (Qiagen, Valencia, CA, USA). Samples were run in dupli-
ognition and remembering foods they ate during the last cate and only samples with a coefficient of variation less
week. Questionnaires were processed by NutritionQuest, than 5% were used in the final analysis. Epitect (Qiagen,
and the resulting data were analyzed at the University of Valencia, CA, USA) bisulfite treated controls, which
Montana. include a methylated and unmethylated human genome
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 4 of 12

sample, along with a no template control were used on buccal cells, PAQLQ and spirometry and therefore, only
each plate. these ‘complete’ visits were considered in the analysis.
Approximately 63% of subcohort, or 20 participants, had
Statistical analysis both a year one and year two ‘complete’ visit, while 12
All analyses were conducted using SAS v9.4 (Cary, NC, participants only had one ‘complete’ visit, which occurred
USA). To evaluate if cross sectional measurements of in either year one or year two, for a total of 52 observa-
IFN CpG sites are correlated with each other and/or tions. Reasons for these 12 participants having one rather
correlated with LINE1 global methylation we estimated than 2  years of data included missing data, participant
Pearson correlation coefficients using the first available not available during scheduled visit, or the participant
observation for each participant (n  =  32). A subset of chose to only participate in one year of the study. Moreo-
17 macro- and micronutrients from the total 73 nutrient ver, in the final GEE models, which were adjusted for sev-
variables generated by the FFQ were chosen after a litera- eral covariates, one participant (two observations) was
ture review of diet as it relates to asthma. Relationships excluded because income and education data was miss-
between a priori selected dietary nutrients and both epi- ing, therefore the results from these models include 50
genetic markers and asthma outcomes were considered observations from 31 participants. Ages ranged from 8 to
in separate models using all available and complete data, 17  years and 47% were male (Table  1). The study popu-
which included multiple visits for some participants. lation was 94% non-Hispanic white. The mean (sd) BMI
Their associations with continuous epigenetic markers percentile was 70.6 (20.1) and 34% (n =  11) were above
(i.e. global and gene-specific methylation) and asthma the 85th percentile, which is considered overweight
measures were evaluated using generalized estimating according to the CDC [34]. Baseline asthma-related res-
equations (GEE), which account for correlations between piratory health values can be found in Table  1. Mean
repeated measures on the same participant. Tertiles of values for both dPFV and FEV1 were at the approximate
dietary nutrients were included in analyses as three-level threshold used to designate poor asthma control [41].
indicator variables to investigate potentially non-linear Mean (sd) LINE-1 methylation was 65.3% (3.4) with a
relationships with epigenetic and asthma outcomes. range of 56.1–73.2%. Mean (sd) IFNγ CpG-54 was 79.6%
Analyses were adjusted for age (continuous) and gender. (4.5) with a range of 68.6–92.4%. Mean (sd) IFNγ CpG-
Although this diet and epigenetics repeated measures 186 was 70.1% (6.6) with a range of 49.1–81.6%. IFNγ
study was not directly related to the indoor air quality CpG-54 and IFNγ CpG-186 observations were moder-
intervention study, we included in our models indica- ately correlated with each other (r  =  0.42; p  =  0.02) as
tors for pre- versus post-intervention winter and home were IFNγ CpG-54 and LINE-1 methylation (r  =  0.44;
intervention assignment (i.e., placebo versus air filter). p = 0.01). IFNγ CpG-186 and LINE-1 methylation were
Inclusion of the following potential confounders: pres- not significantly correlated (r = 0.26; p = 0.15).
ence of cat or dog in home (yes or no), family income
(above or below $50,000) and parent education (col- Table 1  Selected characteristics of subset of ARTIS partici-
lege degree or no college degree) appreciably impacted pants included in epigenetic study
parameter estimates. Therefore, the final model included N Mean SD Range
age, gender, winter and intervention group assignment,
Age 32 12.8 2.5 8.0–17.0
presence of cat or dog, income and education. We inves-
Gender
tigated relationships between epigenetic markers and
 Male 15 (46%)
asthma measures in a similar manner. Due to the num-
 Female 17 (53%)
ber of comparisons (n = 264), a false discovery rate cor-
Ethnicity
rection [40] was applied and adjusted p-values (q-values)
 Non-Hispanic 30 (94%)
were calculated for each relationship where the GEE
BMI percentile 32 70.6 24.1 5.8–99.0
model was used. A threshold for significance was set at
dPFV 32 20.0 14.0 2.0–66.0
q < 0.20 which means we accept that 20% of the observed
% Predicted morning FEV1 32 81.7 19.7 26.9–112.9
significant relationships (i.e. 3.4 out of 17) could be false
% Predicted evening FEV1 32 82.3 19.3 19.3–110.1
positives.
PAQLQ 32 5.6 1.1 3.1–7.0
LINE-1 (%) 32 65.3 3.4 56.1–73.2
Results
A subset of 32 children from the ARTIS cohort partici- IFNγ-54 (%) 32 79.6 4.5 68.6–92.4
pated in this study of diet, asthma health and epigenet- IFNγ-186 (%) 32 70.1 6.6 49.1–81.6
ics and was included in the analyses described here. Diet SD standard deviation, BMI body mass index, dPFV evening to morning peak
flow variability, FEV1 forced expiratory volume in 1 s, PAQLQ pediatric asthma
data was collected once per winter in conjunction with quality of life questionnaire, IFNγ interferon gamma
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 5 of 12

Evaluating dietary nutrients with respect to respiratory Evaluating DNA methylation with respect to respiratory
health health
When considered across categories of calculated intake, We investigated the relationships between methylation
most dietary nutrients failed to show a consistent asso- markers and asthma-related respiratory health meas-
ciation with respiratory health measures, but several urements (Table  4). A one-percentage point increase of
differences in PAQLQ scores were observed between LINE-1 methylation was associated with a 1.24 percent-
participants in the highest third versus the lowest third of age point (95% CI 0.31, 2.16; q = 0.16) increase in dPFV.
intake for some nutrients (Table 2). Phosphatidylcholine Neither IFNγ CpG-54 nor-186 methylation was associ-
was the only selected nutrient that was associated with ated with the respiratory health measures evaluated in
any of the three pulmonary function measure assessed. this study.
Children in the middle tertile relative to the lowest had
16.04% point (95% CI 3.31, 28.78; q = 0.16) higher % pre- Discussion
dicted evening FEV1. Intake of selenium and folate was In this study of dietary nutrients, DNA methylation and
associated with better PAQLQ scores. Specifically, par- asthma-related respiratory health outcomes, we observed
ticipants with the highest tertile of selenium and folate positive associations between several nutrients related to
intake had 1.4 unit (95% CI 0.90, 1.91; q = 0. 01) and 0.92 one-carbon metabolism (e.g. folate, phosphocholine and
unit (95% CI 0.31, 1.53; q = 0.11) higher PAQLQ scores, betaine) and the PAQLQ score. These nutrients were not
respectively. Additionally, nutrients in the one-carbon similarly associated with better self-monitored spirom-
metabolism cycle, phosphocholine (1.11 unit higher etry outcomes, suggesting that the nutrients may posi-
PAQLQ score; 95% CI 0.23, 1.98; q  =  0.16) and betaine tively influence asthma quality of life through another
(0.98 unit higher PAQLQ score; 95% CI 0.30, 1.66; q = 0. mechanism. The composite PAQLQ score is comprised
13) were positively associated with PAQLQ. of symptom, activity and emotion domains; however, a
post hoc analysis, which substituted individual domains
Evaluating dietary nutrients with respect to methylation for the composite PAQLQ in a model with dietary nutri-
outcomes ents (i.e. those nutrients that had significant relationships
Intake of several nutrients was associated with LINE-1 with composite PAQLQ), revealed no difference for the
methylation and methylation at CpG promoter site impact of dietary intake on individual domains. Further-
IFNγ-186, but not for IFNγ-54 (Table 3). Children in the more, the individual domains were highly correlated with
highest tertile of kilocalories (3.2% points higher meth- one another (data not shown) and therefore it is unclear
ylation; 95% CI 0.82, 5.58; q = 0.16) or the middle tertile which of these domains may be more affected by diet.
of protein (2.67% points higher methylation; 95% CI 0.62, Folate (or folic acid) is one of the most prominently
4.71; q = 0.16) had higher LINE-1 methylation. Similarly, studied methyl donors and is known to play a role in
those in the highest tertile of methyl donating nutrients allergic asthma [42]. Many studies have investigated the
free choline (2.18% points higher methylation; 95% CI efficacy of methyl donating nutrient supplementation
0.54, 3.82; q  =  0.16), total choline (2.60% points higher to reduce the risk of asthma development. To date the
methylation; 95% CI 0.60, 4.60; q  =  0.16) and folate results have been inconclusive (see reviews [2, 43]). Many
(4.29% points higher methylation; 95% CI 2.25, 6.34; of these supplementation studies have been either lim-
q  =  0.01) also had higher LINE-1 methylation. Intake ited by ethical concerns or underpowered. We observed
within the middle tertile of kilocalories (4.56% points that folate intake was associated with a higher PAQLQ
lower methylation; 95% CI −7.44, −1.69; q  =  0.09) and score in children with asthma. Based on the most recent
folate (4.05% points lower methylation; 95% CI −6.18, reviews, the evidence suggests that early life exposure to
−1.19; q  =  0.02) compared to the lowest was associ- folate has no major effects on asthma outcomes later in
ated with less IFNγ-186 methylation. However, intake life [44]. However, relevant to our findings, few studies
within middle tertile of monosaturated fat intake (6.88% have investigated the relationship between folate intake
points higher methylation; 95% CI 3.11, 10.62; q = 0.02) and asthma measures in adolescent children with estab-
was associated with more IFNγ-186 methylation. Chil- lished asthma [45]. Folate deficiency has been associated
dren in the highest tertile of betaine intake (4.34% points with asthma-related symptoms and exacerbations [46–
lower methylation; 95% CI −7.25, −1.42; q  =  0.12) and 48]. Folate and betaine are involved in DNA methyla-
both the middle and highest tertile of vitamin B6 intake tion through the formation of S-adenosylmethionine and
(6.57% points lower methylation; 95% CI −10.65, −2.48; homocysteine metabolism and therefore have the poten-
q  =  0.09 and 6.63% points lower methylation; 95% CI tial to affect gene expression, thereby influencing asthma
−11.13, −2.14; q  =  0.12, respectively) had less IFNγ pathogenesis [43]. In addition to methyl donors, selenium
CpG-186 methylation. was also positively associated with PAQLQ score. Fabian
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 6 of 12

Table 2  The relationship between  select nutrients and  asthma health measures by  dietary tertiles in  ARTIS where  the
lowest tertile of intake (T1) is the reference group
Asthma health dPFV % Predicted morning FEV1 % Predicted evening FEV1 PAQLQ
measure
Dietary factor Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value

Kilocalories
 T3:T1 −3.18 (−11.41, 5.06) 0.80 13.09 (−4.04, 30.22) 0.61 4.18 (−14.08, 22.44) 0.87 0.62 (−0.25, 1.48) 0.68
 T2:T1 −0.50 (−8.78, 7.78) 0.98 0.83 (−13.94, 15.60) 0.98 7.04 (−8.31, 22.40) 0.76 −0.34 (−1.20, 0.51) 0.79
Protein
 T3:T1 −6.96 (−20.40, 6.48) 0.72 19.26 (0.48, 38.05) 0.33 10.96 (−2.58, 24.51) 0.55 0.70 (−0.16, 1.56) 0.55
 T2:T1 −3.42 (−14.56, 7.73) 0.83 6.31 (−8.30, 20.92) 0.77 1.14 (−12.42, 14.70) 0.97 0.59 (−0.09, 1.27) 0.51
M-fats
 T3:T1 −2.87 (−10.62, 4.87) 0.81 11.64 (−6.82, 30.11) 0.71 8.55 (−10.72, 27.81) 0.77 0.39 (−0.31, 1.09) 0.71
 T2:T1 −0.57 (−8.81, 7.67) 0.98 3.89 (−9.58, 17.36) 0.83 3.26 (−9.25, 15.77) 0.85 0.52 (−0.03, 1.07) 0.41
S-fats
 T3:T1 −3.99 (−15.87, 7.89) 0.83 16.85 (1.36, 32.35) 0.29 11.73 (0.65, 22.82) 0.33 −0.16 (−0.82, 0.50) 0.87
 T2:T1 −6.56 (−16.72, 3.59) 0.70 16.67 (2.96, 30.37) 0.28 10.19 (−0.41, 20.80) 0.41 −0.05 (−0.77, 0.67) 0.98
Omega 3:6 ratio
 T3:T1 −1.25 (−10.54, 8.03) 0.92 2.42 (−11.59, 16.44) 0.90 −0.39 (−13.84, 13.07) 1.00 −0.13 (−1.18, 0.93) 0.92
 T2:T1 −1.29 (−12.09, 9.51) 0.92 5.40 (−4.00, 14.80) 0.71 1.46 (−6.18, 9.10) 0.90 0.11 (−0.54, 0.75) 0.90
Selenium
 T3:T1 5.44 (−2.98, 13.86) 0.70 6.63 (−6.31, 19.56) 0.73 6.37 (−6.57, 19.30) 0.74 1.40 (0.90, 1.91) 0.01*
 T2:T1 2.52 (−4.56, 9.59) 0.82 0.20 (−14.11, 14.51) 1.00 2.91 (−12.96, 18.77) 0.90 −0.56 (−1.59, 0.47) 0.71
Fiber
 T3:T1 −0.39 (−9.02, 8.25) 0.99 7.75 (−5.27, 20.78) 0.71 5.86 (−8.02, 19.74) 0.77 0.66 (−0.52, 1.84) 0.71
 T2:T1 0.63 (−9.85, 11.11) 0.98 −5.36 (−17.98, 7.27) 0.77 −3.60 (−15.79, 8.59) 0.83 0.32 (−0.38, 1.02) 0.76
Folate
 T3:T1 −3.43 (−14.76, 7.89) 0.83 10.07 (−7.95, 28.09) 0.71 3.75 (−10.53, 18.02) 0.85 0.92 (0.31, 1.53) 0.11*
 T2:T1 −3.03 (−14.23, 8.17) 0.85 −0.03 (−17.50, 17.44) 1.00 −0.24 (−13.99, 13.50) 1.00 −0.25 (−0.83, 0.35) 0.78
Methionine
 T3:T1 −4.83 (−13.90, 4.24) 0.72 14.81 (−0.25, 29.88) 0.36 11.07 (−4.02, 26.17) 0.65 0.52 (−0.37, 1.41) 0.71
 T2:T1 1.87 (−7.53, 11.28) 0.90 5.22 (−12.12, 22.57) 0.83 7.29 (−10.16, 24.73) 0.77 −0.06 (−1.58, 1.46) 0.99
Free choline
 T3:T1 2.44 (−4.68, 9.56) 0.83 11.20 (−6.45, 28.86) 0.70 7.69 (−10.98, 26.36) 0.78 0.00 (−1.34, 1.34) 1.00
 T2:T1 5.35 (−3.47, 14.17) 0.71 −2.64 (14.74, 9.47) 0.88 −0.21 (−13.39, 12.97) 1.00 −0.31 (−1.26, 0.64) 0.83
Glycpp-choline
 T3:T1 2.84 (−5.75, 11.42) 0.83 −3.05 (−18.17, 12.07) 0.89 −3.64 (−19.88, 12.61) 0.88 0.61 (−0.17, 1.39) 0.59
 T2:T1 −1.81 (−9.22, 5.59) 0.87 2.88 (−9.04, 14.80) 0.87 3.70 (−8.10, 15.50) 0.83 0.12 (−0.41, 0.65) 0.87
Pp-choline
 T3:T1 4.54 (−3.07, 12.15) 0.71 −2.95 (−12.99, 7.08) 0.83 −5.21 (−15.57, 5.14) 0.73 1.11 (0.23, 1.98) 0.16*
 T2:T1 −1.03 (−7.55, 5.50) 0.90 −10.81 (−27.27, 5.64) 0.69 −12.35 (−29.79, 5.09) 0.68 0.50 (−0.61, 1.60) 0.77
Ppt-choline
 T3:T1 0.36 (−6.86, 7.58) 0.98 11.60 (−1.45, 24.65) 0.48 −1.13 (−19.21, 16.94) 0.98 0.24 (−0.86, 1.35) 0.88
 T2:T1 5.71 (−2.92, 14.33) 0.69 −3.17 (−15.51, 9.17) 0.85 16.04 (3.31, 28.78) 0.16* −0.19 (−0.58, 0.21) 0.76
Total choline
 T3:T1 1.33 (−6.50, 9.16) 0.90 8.61 (−7.92, 25.14) 0.72 5.96 (−12.23, 24.15) 0.83 0.18 (−1.26, 1.61) 0.92
 T2:T1 3.76 (−4.42, 11.94) 0.76 −6.68 (−20.25, 6.90) 0.74 −2.06 (−16.74, 12.62) 0.92 −0.38 (−1.35, 0.59) 0.80
Betaine
 T3:T1 −2.62 (−9.35, 4.10) 0.80 12.85 (0.98, 24.72) 0.29 9.13 (−1.59, 19.85) 0.51 0.98 (0.30, 1.66) 0.13*
 T2:T1 4.20 (−6.18, 14.58) 0.79 −9.34 (−21.55, 2.86) 0.61 −9.10 (−19.78, 3.59) 0.68 0.21 (−0.56, 0.98) 0.85
Vitamin B-2
 T3:T1 −0.95 (−8.43, 6.54) 0.92 −4.74 (−18.10, 8.61) 0.82 3.81 (−10.97, 18.58) 0.85 0.40 (−0.66, 1.46) 0.80
 T2:T1 1.66 (−8.13, 11.44) 0.90 8.98 (−7.57, 25.52) 0.71 −4.04 (−17.65, 9.56) 0.83 −0.12 (1.12, 0.88) 0.92
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 7 of 12

Table 2  continued
Asthma health dPFV % Predicted morning FEV1 % Predicted evening FEV1 PAQLQ
measure
Dietary factor Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value

Vitamin B-6
 T3:T1 2.53 (−5.37, 10.43) 0.83 4.19 (−10.21, 18.59) 0.83 0.26 (−13.43, 13.94) 1.00 0.46 (−0.21, 1.13) 0.69
 T2:T1 4.64 (−3.10, 12.38) 0.71 −4.96 (−20.50, 10.57) 0.83 0.81 (−14.85, 16.46) 0.98 −0.65 (−1.20, 0.00) 0.36
Vitamin B-12
 T3:T1 −0.02 (−8.26, 8.22) 1.00 6.42 (−8.93, 21.77) 0.77 1.70 (−11.74, 15.13) 0.92 0.79 (0.09, 1.50) 0.29
 T2:T1 1.53 (−7.17, 10.23) 0.90 −2.40 (−16.46, 11.66) 0.90 −3.38 (−16.85, 10.10) 0.86 0.29 (−0.53, 1.11) 0.82

M-fats monosaturated fats, S-fats saturated fats, Glycpp-choline glycerophosphocholine, Pp-choline phosphocholine, Ppt-choline phosphotidylcholine, dPFV evening to
morning peak flow variability, FEV1 forced expiratory volume in 1 s, PAQLQ pediatric asthma quality of life questionnaire
* q-value < 0.20

et  al. found that children with asthma compared to informative, is difficult to interpret in the context of res-
healthy control children had lower plasma levels of sele- piratory health and may be even more complex in this
nium and higher exhaled nitric oxide, a marker of poor cohort of children with asthma. Few studies have looked
lung health [49]. However, a group of Swedish research- at buccal DNA LINE-1 global methylation in healthy
ers found no impact of selenium intake on allergic disease children. A study of 57 healthy girls aged from 6 to 15
in young children [50]. The inconclusive results regarding investigated LINE-1 global methylation in saliva samples
the impact of selenium intake on allergic asthma could and the average (SD) was 75.2 (3.4) [55]. The cells col-
be attributed to the fact that while selenium does have lected from buccal and saliva should be similar, however
antioxidant properties it also has the ability to upregulate the mean LINE-1 methylation in our study overall was
some immune responses [17, 51]. In our analysis sele- considerably lower, which could be attributed to asthma
nium status is associated with better asthma quality of status. It is clear from the literature that intake of methyl
life measures, however, this nutrient was not associated donors can result in measureable changes to the mam-
with LINE-1 or IFNγ methylation profiles. malian epigenome [20]. Our study shows that dietary
Among the dietary nutrients investigated in this study intake of folate, free choline, and total choline is posi-
only phosphatidylcholine was modestly associated tively associated with LINE-1 methylation. By conven-
with self-administered spirometry measures, specifi- tion, an increase in global methylation is thought to be
cally higher evening FEV1. Phosphatidylcholine is phos- protective, while a shift toward genome-wide hypometh-
pholipid and a major dietary source of choline, which is ylation is often associated with a poor health outcome
involved in one-carbon metabolism. Phospholipids can or disease [56, 57]. Nevertheless, our data suggested that
also impact T cell function in a number of ways includ- global methylation was positively associated with dPFV,
ing membrane fluidity and gene expression, which could an indicator of airway hyper-reactivity. This finding
have indirect immunomodulatory effects [52]. There- could be a characteristic of the study population, which
fore, our observation of a positive association between had relatively low average global methylation. Further,
phosphatidylcholine and FEV1 in these children could DNA methylation is dynamic and global methylation is
be reflective of reduced lung inflammation. However, a reflection of the epigenetic changes occurring at many
this association was not consistent across tertiles of gene locations.
phosphatidylcholine intake nor was there a consistent Our study specifically focused on IFNγ as a candidate
response across the different FEV1 measures, evening gene, hypothesizing that this gene would lie in the mech-
versus morning. These concerns together with the stud- anistic pathway linking dietary intake to asthma health
ies that have linked phosphatidylcholine to cardiovascu- outcomes in children. Previous studies have established
lar disease [53] and related inflammatory symptoms [54], that IFNγ CpG-54 and -186 (-53 and -190 are the cor-
suggest that the association between phosphatidylcholine responding murine CpGs) are relevant to allergic out-
intake and FEV1 should be interpreted with caution. comes in animal models [58, 59] and humans [60–62].
In addition to evaluating the relationship between In the mouse, Jones et  al. showed that these CpGs are
nutritional intake and asthma health, a potential epige- functionally relevant (i.e., methylation status affects tran-
netic mechanism was examined by determining if global scription of the IFNγ gene) and that de novo methyla-
methylation or IFNγ promoter methylation was associ- tion of these sites plays a key role in Th2 polarization at
ated with nutritional intake. Global methylation, though least within the CD4+ T cells [63]. In an human asthma
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 8 of 12

Table 3  The relationship between select nutrients and DNA methylation markers by dietary tertiles in ARTIS, where the
lowest tertile of intake (T1) is the reference group
Epigenetic measure IFNγ CpG-54 IFNγ CpG-186 LINE-1
Dietary factor Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value

Kilocalories
 T3:T1 0.46 (−2.46, 3.38) 0.90 −0.11 (−4.35, 4.13) 1.00 3.20 (0.82, 5.58) 0.16*
 T2:T1 −0.40 (−2.87, 2.07) 0.90 −4.56 (−7.44, −1.69) 0.09* 0.00 (−2.45, 2.46) 1.00
Protein
 T3:T1 2.60 (−0.90, 6.10) 0.65 −0.04 (−3.41, 3.32) 1.00 1.62 (−0.79, 4.03) 0.69
 T2:T1 2.17 (−0.46, 4.80) 0.55 2.02 (−1.24, 5.27) 0.71 2.67 (0.62, 4.71) 0.16*
M-fats
 T3:T1 −1.10 (−3.70, 1.51) 0.77 4.77 (−0.04, 9.57) 0.36 1.14 (−1.23, 3.51) 0.76
 T2:T1 −2.42 (−4.74, −0.11) 0.33 6.86 (3.11, 10.62) 0.02* 1.12 (−1.00, 3.24) 0.72
S-fats
T3:T1 −0.75 (−3.11, 1.61) 0.83 5.11 (−1.08, 11.31) 0.55 1.52 (−1.09, 4.13) 0.71
T2:T1 1.02 (−1.91, 3.96) 0.82 3.00 (−2.48, 8.48) 0.71 1.26 (−1.45, 3.97) 0.76
Omega 3:6 ratio
 T3:T1 1.44 (−1.23, 4.10) 0.71 2.29 (−1.85, 6.44) 0.71 −0.09 (−2.70, 2.52) 0.99
 T2:T1 3.72 (0.44, 7.01) 0.29 0.24 (−4.72, 5.20) 0.98 0.96 (−1.03, 2.95) 0.74
Selenium
 T3:T1 0.34 (−2.45, 3.13) 0.92 −1.40 (−5.17, 2.38) 0.81 2.32 (−0.15, 4.79) 0.45
 T2:T1 1.40 (−0.60, 3.40) 0.68 −2.38 (−5.01, 0.24) 0.45 −0.18 (−2.69, 2.33) 0.98
Fiber
 T3:T1 0.53 (−2.31, 3.37) 0.90 −4.67 (−9.09, −0.24) 0.33 1.89 (−1.02, 4.80) 0.69
 T2:T1 0.19 (−2.29, 2.68) 0.98 −2.52 (−6.54, 1.49) 0.71 −0.27 (−2.70, 2.16) 0.94
Folate
 T3:T1 2.93 (0.06, 5.80) 0.36 2.46 (−2.25, 7.17) 0.72 4.29 (2.25, 6.34) 0.01*
 T2:T1 1.95 (−0.03, 3.93) 0.36 −4.05 (−6.18, −1.91) 0.02* 0.46 (−1.24, 2.16) 0.85
Methionine
 T3:T1 0.63 (−2.42, 3.68) 0.89 −2.70 (−6.86, 1.45) 0.69 1.12 (−1.78, 4.03) 0.80
 T2:T1 0.43 (−2.25, 3.11) 0.90 −1.38 (−5.68, 2.92) 0.83 1.58 (−1.04, 4.21) 0.71
Free choline
 T3:T1 1.33 (−1.08, 3.74) 0.71 −2.61 (−5.84, 0.63) 0.55 2.18 (0.54, 3.82) 0.16*
 T2:T1 1.29 (−1.03, 3.60) 0.71 −3.28 (−6.33, −0.24) 0.29 0.01 (−1.82, 1.85) 1.00
Glycpp-choline
 T3:T1 0.44 (−2.23, 3.12) 0.90 −2.81 (−5.95, 0.33) 0.48 0.66 (−1.48, 2.80) 0.83
 T2:T1 1.68 (−0.54, 3.89) 0.65 −0.31 (−3.84, 3.22) 0.97 0.24 (−1.74, 2.21) 0.92
Pp-choline
 T3:T1 0.50 (−2.65, 3.65) 0.90 −1.77 (−5.27, 1.73) 0.73 0.41 (−1.43, 2.25) 0.88
 T2:T1 1.47 (−0.52, 3.46) 0.65 −1.54 (−5.83, 2.75) 0.82 −2.08 (−4.41, 0.24) 0.48
Ppt-choline
 T3:T1 1.49 (−1.08, 4.06) 0.71 −2.33 (−6.06, 1.39) 0.71 0.64 (−1.66, 2.94) 0.84
 T2:T1 −0.78 (−3.40, 1.84) 0.83 −1.55 (−6.86, 3.76) 0.83 −1.66 (−4.03, 0.71) 0.68
Total choline
 T3:T1 1.29 (−1.06, 3.64) 0.71 −3.47 (−6.68, −0.26) 0.29 2.60 (0.60, 4.60) 0.16*
 T2:T1 0.85 (−1.41, 3.11) 0.80 −3.58 (−6.59, −0.58) 0.28 0.42 (−1.88, 2.73) 0.90
Betaine
 T3:T1 −0.00 (−2.50, 2.49) 1.00 −4.34 (−7.25, −1.42) 0.12* 1.18 (−1.25, 3.62) 0.74
 T2:T1 1.52 (−1.21, 4.26) 0.71 1.28 (−4.28, 1.73) 0.77 1.65 (−0.61, 3.91) 0.65
Vitamin B-2
 T3:T1 0.62 (−2.44, 3.68) 0.89 −4.19 (−7.91, −0.48) 0.29 2.47 (−0.37, 5.32) 0.51
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 9 of 12

Table 3  continued
Epigenetic measure IFNγ CpG-54 IFNγ CpG-186 LINE-1
Dietary factor Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value

 T2:T1 1.41 (−0.74, 3.56) 0.69 −4.03 (−7.86, −0.20) 0.33 0.23 (−2.48, 2.94) 0.97
Vitamin B-6
 T3:T1 2.05 (−1.04, 5.14) 0.69 −6.63 (−11.13, −2.14) 0.12* 1.44 (−1.65, 4.52) 0.76
 T2:T1 0.66 (−1.80, 3.12) 0.85 −6.57 (−10.65, −2.48) 0.09* 0.13 (−2.39, 2.65) 0.98
Vitamin B-12
 T3:T1 1.12 (−2.31, 4.55) 0.83 −2.31 (−7.00, 2.39) 0.74 0.59 (−2.43, 3.60) 0.90
 T2:T1 2.21 (−0.48, 4.91) 0.55 1.84 (−2.37, 6.05) 0.77 0.87 (−2.09, 3.82) 0.83
M-fats monosaturated fats, S-fats saturated fats, Glycpp-choline glycerophosphocholine, Pp-choline phosphocholine, Ppt-choline phosphotidylcholine, IFNγ interferon
gamma
* q-value <0.20

Table 4  The relationship between epigenetic measurements and asthma health outcomes in ARTIS


Epigenetic marker dPFV % Predicted morning FEV1 % Predicted evening FEV1 PAQLQ
Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value Estimate (95% CI) q-value

LINE-1 1.24 (0.31, 2.16) 0.16* −0.89 (−3.18, 1.41) 0.80 −1.19 (−3.41, 1.03) 0.72 0.04 (−0.11, 0.20) 0.84
IFNγ CpG-186 0.58 (0.06, 1.09) 0.29 −0.53 (−1.79, 0.72) 0.77 −0.57 (−1.62, 0.48) 0.72 0.03 (0.00, 0.07) 0.44
IFNγ CpG-54 0.22 (−0.74, 1.19) 0.87 −0.57 (−1.92, 0.77) 0.77 −0.84 (−2.10, 0.42) 0.69 0.02 (−0.07, 0.11) 0.85
dPFV evening to morning peak flow variability, FEV1 forced expiratory volume in 1 s, PAQLQ pediatric asthma quality of life questionnaire, IFNγ interferon gamma
* q-value <0.20

cohort, Lovinsky-Desir et al. showed that there are differ- a dietary nutrient and methylation at this site could
ential methylation profiles for these CpGs relative to age, impact the Th1/Th2 balance by increasing the expres-
sex and tissue type [64]. For example, when methylation sion of IFNγ. Overall nutrient intake has previously been
profiles of buccal cells and CD4+  lymphocytes isolated linked to IFNγ production [65]. However, overnutrition
from whole blood were compared, IFNγ CpG-186 was is not likely a preferable or effective asthma interven-
correlated for males but not females. Further, methyla- tion especially due to the potential links between obe-
tion values for IFNγ CpG-54 and -186 were correlated for sity, inflammation, and asthma. Monosaturated fat was
children and adults in CD4+  lymphocytes but only for the only nutrient we found to be positively associated
adults in buccal cells. White et al. investigated IFNγ pro- with IFNγ-186 methylation. In a study of approximately
moter methylation profiles by in vitro polyclonal expan- 1200 adolescent children conducted in Taiwan, intake
sion of CD4+ and CD8+ T cells sorted from peripheral of monosaturated fats was inversely associated with risk
blood mononuclear cells [62]. When samples collected of asthma [66]. By contrast, a study of nearly 4000 adult
from adolescent children were stratified by atopic sta- European participants found that intake of monosatu-
tus, the authors found that, for CD8+ T cells under Th1 rated fats was positively associated with allergic sensiti-
polarizing conditions, IFNγ CpG sites -54 and -186 were zation [67].
less methylated in the non-atopic children. IFNγ CpG promoter methylation at site -54 and -186
When evaluating the impact of diet on IFNγ promoter was not associated with respiratory health measures
methylation, we found that only IFNγ CpG-186 meth- or PAQLQ. This suggests that the positive relationship
ylation patterns were affected by selected nutrients. We revealed between PAQLQ composite score and intake of
observed that intake of kilocalories and three methyl selenium, folate, phosphocholine, and betaine may not
donating nutrients was associated with less IFNγ CpG- be working directly through epigenetic modification of
186 methylation, while children who had higher intake these specific sites as we had hypothesized.
of monosaturated fats had more IFNγ CpG-186 meth-
ylation. Based on the functional data available for this Limitations and cautions
CpG site, which we note does not come from buccal We evaluated several dietary macro- and micronutri-
cells, we speculate that a negative association between ents in this study, but these factors likely include only a
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 10 of 12

portion of the exogenous factors that could influence generalized estimating equations; glycpp-choline: glycerophosphocholine;
IFNγ: interferon gamma; LINE-1: long interspersed nuclear elements; M-fats:
DNA methylation in this population. While FFQs are monosaturated fats; NO2: nitrogen dioxide; PAQLQ: pediatric asthma quality of
an accepted and validated method for acquiring per- life questionnaire; PCR: polymerase chain reaction; PEF: peak expiratory flow;
sonal dietary information, we note that the portion sizes PM: particulate matter; PM2.5: fine PM or particles less than 2.5 micrometers in
aerodynamic diameter; Pp-choline: phosphocholine; Ppt-choline: phosphoti-
and specific foodstuffs were self-reported by the partici- dylcholine; Runx3: runt-related transcription factor 3; S-fats: saturated fats; SD:
pants with assistance and input from parents. Though standard deviation; Tregs: regulatory T cells; Th1: type 1 T helper; Th2: type 2 T
it is widely accepted, BMI may actually be a poor indi- helper; USEPA: U.S. Environmental Protection Agency.
cator for obesity in children and adolescents who have Authors’ contributions
large, lean body mass from physical activity, high mus- LM aided in the design of the study, acquisition and analysis of data, and with
cularity, or frame size. By focusing on select candidate drafting the manuscript. TW aided in the design of the study and with drafting
the manuscript. ES contributed to the data analysis strategy and interpretation
DNA methylation markers, we recognize that numerous as well as with drafting the manuscript. YHC provided support for epigenetic
inflammatory pathways involving diet and asthma may assays and with manuscript revisions. BB aided in the design, collection, and
not have been captured. We also are limited in our inter- analysis of diet related data as well as manuscript revisions. CN aided in the
design of the study, data analysis strategy and interpretation, and with draft-
pretation of the DNA methylation data because we did ing the manuscript. All authors read and approved the final manuscript.
not measure IFNγ expression or protein levels in these
samples. For example, we found some dietary factors to Author details
1
 School of Public Health, University of Michigan, 1420 Washington Heights,
be negatively associated with DNA methylation, which Ann Arbor, MI 48109, USA. 2 Center for Environmental Health Sciences, Uni-
could be informative for asthma intervention strategies, versity of Montana, 32 Campus Drive‑159 Skaggs, Missoula, MT 59812, USA.
but such interpretations require further assessment as 3
 Department of Health and Human Performance, University of Montana, 32
Campus Drive, Missoula, MT 59812, USA.
methylation changes do not necessarily translate to func-
tional changes in the target tissue. Finally, although we Acknowledgements
accounted for false discoveries, we recognize that several The authors would like to thank Dr. Jackie Goodrich and Virginia Kay for
providing support for epigenetic assay development, Emily Weiler for field col-
statistical tests were performed and would expect some lection of data, Dr. Dana Dolinoy for mentorship and Carolyn Hester for aiding
significant results due to chance alone. Thus these obser- in study management and organization.
vations should be considered exploratory and requiring
Competing interests
of further study in other populations. The authors declare that they have no competing interests.

Conclusions Availability of data and materials


Please contact author for data requests.
Within this cohort of childhood asthmatics, we sought
to identify dietary nutrients that may be beneficial for Ethics approval and consent to participate
respiratory health. In addition we measured LINE-1 The study was approved by the University of Montana Institutional Review
Board (protocol numbers 233_10 and 152_11). In addition to the informed
and IFNγ (CpG-54 and -186) methylation levels to iden- consent procedures for the parent study, children were separately assented
tify pathways whereby diet influences the health among to participate in this diet and epigenetics study and parents signed a parental
children with asthma. In this study, selenium and several permission and informed consent form.
nutrients involved in the one-carbon metabolism path- Funding
way were associated with improved asthma quality of This research was supported by the National Institute of Environmental Health
life measures. Furthermore, these data showed that some Sciences (1R01ES016336–01 and 3R01ES016336–02S1). Additional support was
provided by a COBRE award (P20RR017670) and the National Institute of Gen-
dietary constituents were associated with both global and eral Medical Sciences (P20GM103474 and 1U54GM104944). LM was partially
gene specific methylation in children with asthma. The supported by an NIH NRSA T32 training program Grant (#E2007062, PI: Rita
two IFNγ CpG sites that were investigated appear to be Loch-Caruso). The content is solely the responsibility of the authors and does
not necessarily represent the official views of the National Institutes of Health.
uniquely affected by intake of micro- and macronutrients.
Received: 8 June 2016 Accepted: 17 February 2017
Additional file

Additional file 1: Table S1. Primers and amplification conditions for


PCR and pyrosequencing experiments. Figure S1. Description of ARTIS
sub-study design. References
1. Akinbami LJ, et al. Trends in asthma prevalence, health care use, and
mortality in the United States, 2001–2010. NCHS Data Brief. 2012;94:1–8.
Abbreviations 2. Han YY, et al. Diet and asthma: vitamins and methyl donors. Lancet Respir
ARTIS: asthma randomized trial of indoor wood smoke; BMI: body mass Med. 2013;1(10):813–22.
index; CDC: U.S. Centers for Disease Control and Prevention; CI: confidence 3. Thuesen BH, et al. Atopy, asthma, and lung function in relation to folate
interval; CpG: regions of DNA where a cytosine nucleotide is followed by a and vitamin B(12) in adults. Allergy. 2010;65(11):1446–54.
guanine nucleotide; DNA: deoxyribonucleic acid; dPFV: diurnal peak flow 4. Mehta AK, et al. Choline attenuates immune inflammation and sup-
variability; FEV1: forced expiratory volume in the first second; FFQ: food presses oxidative stress in patients with asthma. Immunobiology.
frequency questionnaires; FOXp3: transcription factor forkhead box p3; GEE: 2010;215(7):527–34.
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 11 of 12

5. Barros R, et al. Adherence to the Mediterranean diet and fresh fruit intake 33. Semmens EO, et al. Indoor particulate matter in rural, wood stove heated
are associated with improved asthma control. Allergy. 2008;63(7):917–23. homes. Environ Res. 2015;138:93–100.
6. Barros R, et al. Dietary intake of alpha-linolenic acid and low ratio of 34. BMI percentile calculator for child and teen. https://nccd.cdc.gov/
n-6:n-3 PUFA are associated with decreased exhaled NO and improved dnpabmi/calculator.aspx. Accessed 1 Dec 2016.
asthma control. Br J Nutr. 2011;106(3):441–50. 35. Block G, et al. Revision of dietary analysis software for the health habits
7. Castro-Rodriguez JA, et al. Mediterranean diet as a protective factor for and history questionnaire. Am J Epidemiol. 1994;139(12):1190–6.
wheezing in preschool children. J Pediatr. 2008;152(6):823–8. 36. Block G, DiSogra C. WIC dietary assessment validation study. Final report.
8. Chatzi L, et al. Diet, wheeze, and atopy in school children in Menorca, Alexandria: US Department of Agriculture, Food and Nutrition Service;
Spain. Pediatr Allergy Immunol. 2007;18(6):480–5. 1995.
9. Chatzi L, et al. Mediterranean diet in pregnancy is protective for wheeze 37. Block G, et al. Comparison of two dietary questionnaires validated against
and atopy in childhood. Thorax. 2008;63(6):507–13. multiple dietary records collected during a 1-year period. J Am Diet
10. de Batlle J, et al. Mediterranean diet is associated with reduced asthma Assoc. 1992;92:686–93.
and rhinitis in Mexican children. Allergy. 2008;63(10):1310–6. 38. Block G, et al. Validation of a self-administered diet history questionnaire
11. Garcia-Marcos L, et al. Relationship of asthma and rhinoconjunctivitis using multiple diet records. J Clin Epidemiol. 1990;43(12):1327–35.
with obesity, exercise and Mediterranean diet in Spanish schoolchildren. 39. Juniper EF, et al. Measuring quality of life in children with asthma. Qual
Thorax. 2007;62(6):503–8. Life Res. 1996;5(1):35–46.
12. Romieu I, et al. Dietary intake, lung function and airway inflammation 40. Benjamini Y, Hochberg Y. Controlling the false discovery rate—a
in Mexico City school children exposed to air pollutants. Respir Res. practical and powerful approach to multiple testing. J R Stat Soc Series
2009;10:122. B-Methodol. 1995;57(1):289–300.
13. Berthon BS, Wood LG. Nutrition and respiratory health-feature review. 41. Lung National Heart, Institute Blood. Expert panel report 3: guidelines
Nutrients. 2015;7(3):1618–43. for the diagnosis and management of asthma. Bethesda: National Heart
14. Berthon BS, et al. Investigation of the association between dietary Lung, and Blood Institute; 2007.
intake, disease severity and airway inflammation in asthma. Respirology. 42. Sharma S, Litonjua A. Asthma, allergy, and responses to methyl donor
2013;18(3):447–54. supplements and nutrients. J Allergy Clin Immunol. 2014;133(5):1246–54.
15. Cortessis VK, et al. Environmental epigenetics: prospects for studying 43. Blatter J, et al. Folate and asthma. Am J Respir Crit Care Med.
epigenetic mediation of exposure–response relationships. Hum Genet. 2013;188(1):12–7.
2012;131(10):1565–89. 44. Crider KS, et al. Prenatal folic acid and risk of asthma in children: a system-
16. Lovinsky-Desir S, Miller RL. Epigenetics, asthma, and allergic dis- atic review and meta-analysis. Am J Clin Nutr. 2013;98(5):1272–81.
eases: a review of the latest advancements. Curr Allergy Asthma Rep. 45. Lin JH, et al. Relationships between folate and inflammatory features of
2012;12(3):211–20. asthma. J Allergy Clin Immunol. 2013;131(3):918–20.
17. Norton RL, Hoffmann PR. Selenium and asthma. Mol Aspects Med. 46. Matsui EC, Matsui W. Higher serum folate levels are associated
2012;33(1):98–106. with a lower risk of atopy and wheeze. J Allergy Clin Immunol.
18. Bansal P, et al. Intranasal administration of a combination of choline chlo- 2009;123(6):1253–9.
ride, vitamin C, and selenium attenuates the allergic effect in a mouse 47. Farres MN, et al. Study of folate status among Egyptian asthmatics. Intern
model of airway disease. Free Radic Biol Med. 2014;73:358–65. Med. 2011;50(3):205–11.
19. North ML, Ellis AK. The role of epigenetics in the developmental origins of 48. Blatter J, et al. Folate deficiency, atopy, and severe asthma exacerbations
allergic disease. Ann Allergy Asthma Immunol. 2011;106(5):355–61. in Puerto Rican children. Ann Am Thorac Soc. 2016;13(2):223–30.
20. Dolinoy DC. The agouti mouse model: an epigenetic biosensor for nutri- 49. Fabian E, et al. Nutritional supplements and plasma antioxidants in child-
tional and environmental alterations on the fetal epigenome. Nutr Rev. hood asthma. Wien Klin Wochenschr. 2013;125(11–12):309–15.
2008;66(Suppl 1):S7–11. 50. Rosenlund H, et al. Antioxidant intake and allergic disease in children. Clin
21. Hollingsworth JW, et al. In utero supplementation with methyl Exp Allergy. 2012;42(10):1491–500.
donors enhances allergic airway disease in mice. J Clin Invest. 51. Hoffmann PR. Selenium and asthma: a complex relationship. Allergy.
2008;118(10):3462–9. 2008;63(7):854–6.
22. Lloyd CM, Hawrylowicz CM. Regulatory T cells in asthma. Immunity. 52. Pollock AH, et al. Prolonged Intake of dietary lipids alters mem-
2009;31(3):438–49. brane structure and T cell responses in LDLr−/− Mice. J Immunol.
23. Nadeau K, et al. Ambient air pollution impairs regulatory T-cell function in 2016;196(10):3993–4002.
asthma. J Allergy Clin Immunol. 2010;126(4):845–52. 53. Wang Z, et al. Gut flora metabolism of phosphatidylcholine promotes
24. Friso S, et al. One-carbon metabolism and epigenetics. Mol Aspects Med. cardiovascular disease. Nature. 2011;472(7341):57–63.
2016. 54. de Pablo MA, Puertollano MA, Alvarez de Cienfuegos G. Biological and
25. Calderon C, et al. T-cell cytokine profiles are altered in childhood asthma clinical significance of lipids as modulators of immune system functions.
exacerbation. Respirology. 2009;14(2):264–9. Clin Diagn Lab Immunol. 2002;9(5):945–50.
26. Crinnion WJ. Do environmental toxicants contribute to allergy and 55. Wu HC, et al. Correlation of DNA methylation levels in blood and
asthma? Altern Med Rev. 2012;17(1):6–18. saliva DNA in young girls of the LEGACY Girls study. Epigenetics.
27. Smart JM, et al. Polyclonal and allergen-induced cytokine responses in 2014;9(7):929–33.
adults with asthma: resolution of asthma is associated with normalization 56. Rozek LS, et al. Epigenetics: relevance and implications for public health.
of IFN-gamma responses. J Allergy Clin Immunol. 2002;110(3):450–6. Annu Rev Public Health. 2014;35:105–22.
28. Meng H, et al. In vitro production of IL-6 and IFN-gamma is influenced by 57. Perera F, Herbstman J. Prenatal environmental exposures, epigenetics,
dietary variables and predicts upper respiratory tract infection incidence and disease. Reprod Toxicol. 2011;31(3):363–73.
and severity respectively in young adults. Front Immunol. 2015;6:94. 58. Liu J, et al. Combined inhaled diesel exhaust particles and allergen
29. Mohammadi-Shahrokhi V, et al. Immunomodulatory effects of adjuvants exposure alter methylation of T helper genes and IgE production in vivo.
CPG, MPLA, and BCG on the Derp2-induced acute asthma at early life in Toxicol Sci. 2008;102(1):76–81.
an animal model of BALB/c Mice. Inflammation. 2016. 59. Brand S, et al. DNA methylation of TH1/TH2 cytokine genes affects sen-
30. Santeliz JV, et al. Amb a 1-linked CpG oligodeoxynucleotides reverse sitization and progress of experimental asthma. J Allergy Clin Immunol.
established airway hyperresponsiveness in a murine model of asthma. J 2012;29(6):1602–10.
Allergy Clin Immunol. 2002;109(3):455–62. 60. Runyon RS, et al. Asthma discordance in twins is linked to epigenetic
31. Noonan CW, Ward TJ. Asthma randomized trial of indoor wood modifications of T cells. PLoS ONE. 2012;7(11):e48796.
smoke (ARTIS): rationale and methods. Contemp Clin Trials. 61. Kohli A, et al. Secondhand smoke in combination with ambient air
2012;33(5):1080–7. pollution exposure is associated with increasedx CpG methylation and
32. Ward TJ, et al. Efficacy of interventions targeting household air pol- decreased expression of IFN-gamma in T effector cells and Foxp3 in T
lution from residential wood stoves. J Expo Sci Environ Epidemiol. regulatory cells in children. Clin Epigenetics. 2012;4(1):17.
2017;27(1):64–71.
Montrose et al. Allergy Asthma Clin Immunol (2017) 13:12 Page 12 of 12

62. White GP, et al. CpG methylation patterns in the IFNgamma promoter in 65. van den Brink GR, et al. Feed a cold, starve a fever? Clin Diagn Lab Immu-
naive T cells: variations during Th1 and Th2 differentiation and between nol. 2002;9(1):182–3.
atopics and non-atopics. Pediatr Allergy Immunol. 2006;17(8):557–64. 66. Huang SL, Pan WH. Dietary fats and asthma in teenagers: analyses of the
63. Jones B, Chen J. Inhibition of IFN-gamma transcription by site- first Nutrition and Health Survey in Taiwan (NAHSIT). Clin Exp Allergy.
specific methylation during T helper cell development. EMBO J. 2001;31(12):1875–80.
2006;25(11):2443–52. 67. Heinrich J, et al. Allergic sensitization and diet: ecological analysis in
64. Lovinsky-Desir S, et al. DNA methylation of the allergy regulatory gene selected European cities. Eur Respir J. 2001;17(3):395–402.
interferon gamma varies by age, sex, and tissue type in asthmatics. Clin
Epigenetics. 2014;6(1):9.

Submit your next manuscript to BioMed Central


and we will help you at every step:
• We accept pre-submission inquiries
• Our selector tool helps you to find the most relevant journal
• We provide round the clock customer support
• Convenient online submission
• Thorough peer review
• Inclusion in PubMed and all major indexing services
• Maximum visibility for your research

Submit your manuscript at


www.biomedcentral.com/submit

You might also like